Abstract

Abstract Introduction: Of all gynecologic cancers, epithelial ovarian cancer (OCa) stands out with the highest mortality rates. Despite all efforts, 90% of individuals who receive standard surgical and cytotoxic therapy experience disease recurrence. The potential involvement of leukemia inhibitory factor (LIF) and its receptor (LIFR) in the progression of OCa is still obscure. In this study, we examined the mechanisms by which disruption of LIF/LIFR autocrine loops contributes to the cell death of ovarian cancer cells. Methods: The expression profiles of LIF and LIFR were obtained using 24 distinct OCa model cells, comprising both primary and established cell lines. The effects of EC359 on OCa cells were evaluated using reporter assays, colony formation, cell death, and cell viability. Flow cytometry was utilized to analyze lipid peroxidation. The Agilent Seahorse XF Pro Analyzer was used to calculate the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR). Furthermore, flow cytometry was used for the preparation and analysis of lymphocytes. Mechanistic investigations were performed with RT-qPCR and RNA-seq analysis. The efficacy of the LIFR inhibitor EC359 as a targeted therapy was investigated using cell-based xenografts, syngeneic xenografts, patient-derived organoids (PDO), and patient-derived xenograft (PDX) models. Results: Analysis of cancer databases revealed that elevated expression of LIF or LIFR was associated with poor progression-free survival of OCa patients and a predictor of poor response to chemotherapy. Using 24 different primary and established OCa cell lines that represent four subtypes of epithelial OCa, we demonstrated that LIF/LIFR autocrine signaling is active in OCa. Moreover, treatment with EC359, a novel LIFR inhibitor, significantly reduced OCa cell viability and cell survival with an IC50 ranging from 5 to 50 nM. Furthermore, EC359 diminished the stemness of OCa cells. Mechanistic studies using RNA-seq and rescue experiments unveiled that EC359 primarily induced ferroptosis by suppressing the glutathione antioxidant defense system. Using multiple in vitro, ex vivo, and in vivo models including organoids, cell-based xenografts, patient-derived explants, and xenograft tumors, we demonstrated that EC359 dramatically reduced the growth and progression of OCa. Additionally, EC359 therapy considerably improved tumor immunogenicity by robust CD45+ leukocyte tumor infiltration and polarizing tumor-associated macrophages (TAMs) toward M1 phenotype while showing no impact on normal T-, B-, and other immune cells. Conclusions: Collectively, our findings indicate that the LIF/LIFR autocrine loop plays an essential role in OCa progression and that EC359 could be a promising therapeutic agent for OCa. Citation Format: Behnam Ebrahimi, Suryavathi Viswanadhapalli, Uday P. Pratap, Rahul Gopalam, Xue Yang, Prabhakar P. Venkata, Viktor Drel, Bindu Santhamma, Swapna Konda, Xiaonan Li, Alondra L. Rodriguez Sanchez, Hui Yan, Gangadhara R. Sareddy, Zhenming Xu, Brij B. Singh, Philip T. Valente, Yidong Chen, Zhao Lai, Manjeet Rao, Edward R. Kost, Tyler Curiel, Rajeshwar R. Tekmal, Hareesh B. Nair, Ratna K. Vadlamudi. Pharmacological inhibition of the LIF-LIFR autocrine loop reveals vulnerability of ovarian cancer to ferroptosis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 5998.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call